1
|
Elhawary NA, AlJahdali IA, Abumansour IS, Azher ZA, Falemban AH, Madani WM, Alosaimi W, Alghamdi G, Sindi IA. Phenotypic variability to medication management: an update on fragile X syndrome. Hum Genomics 2023; 17:60. [PMID: 37420260 PMCID: PMC10329374 DOI: 10.1186/s40246-023-00507-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
This review discusses the discovery, epidemiology, pathophysiology, genetic etiology, molecular diagnosis, and medication-based management of fragile X syndrome (FXS). It also highlights the syndrome's variable expressivity and common comorbid and overlapping conditions. FXS is an X-linked dominant disorder associated with a wide spectrum of clinical features, including but not limited to intellectual disability, autism spectrum disorder, language deficits, macroorchidism, seizures, and anxiety. Its prevalence in the general population is approximately 1 in 5000-7000 men and 1 in 4000-6000 women worldwide. FXS is associated with the fragile X messenger ribonucleoprotein 1 (FMR1) gene located at locus Xq27.3 and encodes the fragile X messenger ribonucleoprotein (FMRP). Most individuals with FXS have an FMR1 allele with > 200 CGG repeats (full mutation) and hypermethylation of the CpG island proximal to the repeats, which silences the gene's promoter. Some individuals have mosaicism in the size of the CGG repeats or in hypermethylation of the CpG island, both produce some FMRP and give rise to milder cognitive and behavioral deficits than in non-mosaic individuals with FXS. As in several monogenic disorders, modifier genes influence the penetrance of FMR1 mutations and FXS's variable expressivity by regulating the pathophysiological mechanisms related to the syndrome's behavioral features. Although there is no cure for FXS, prenatal molecular diagnostic testing is recommended to facilitate early diagnosis. Pharmacologic agents can reduce some behavioral features of FXS, and researchers are investigating whether gene editing can be used to demethylate the FMR1 promoter region to improve patient outcomes. Moreover, clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 and developed nuclease defective Cas9 (dCas9) strategies have promised options of genome editing in gain-of-function mutations to rewrite new genetic information into a specified DNA site, are also being studied.
Collapse
Affiliation(s)
- Nasser A. Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Imad A. AlJahdali
- Department of Community Medicine, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Iman S. Abumansour
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Zohor A. Azher
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Alaa H. Falemban
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Mecca, 24382 Saudi Arabia
| | - Wefaq M. Madani
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Wafaa Alosaimi
- Department of Hematology, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Ghydda Alghamdi
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Preparatory Year Program, Batterjee Medical College, Jeddah, 21442 Saudi Arabia
| |
Collapse
|
2
|
Miles KD, Doll CA. Chloride imbalance in Fragile X syndrome. Front Neurosci 2022; 16:1008393. [PMID: 36312023 PMCID: PMC9596984 DOI: 10.3389/fnins.2022.1008393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Developmental changes in ionic balance are associated with crucial hallmarks in neural circuit formation, including changes in excitation and inhibition, neurogenesis, and synaptogenesis. Neuronal excitability is largely mediated by ionic concentrations inside and outside of the cell, and chloride (Cl-) ions are highly influential in early neurodevelopmental events. For example, γ-aminobutyric acid (GABA) is the main inhibitory neurotransmitter of the mature central nervous system (CNS). However, during early development GABA can depolarize target neurons, and GABAergic depolarization is implicated in crucial neurodevelopmental processes. This developmental shift of GABAergic neurotransmission from depolarizing to hyperpolarizing output is induced by changes in Cl- gradients, which are generated by the relative expression of Cl- transporters Nkcc1 and Kcc2. Interestingly, the GABA polarity shift is delayed in Fragile X syndrome (FXS) models; FXS is one of the most common heritable neurodevelopmental disorders. The RNA binding protein FMRP, encoded by the gene Fragile X Messenger Ribonucleoprotein-1 (Fmr1) and absent in FXS, appears to regulate chloride transporter expression. This could dramatically influence FXS phenotypes, as the syndrome is hypothesized to be rooted in defects in neural circuit development and imbalanced excitatory/inhibitory (E/I) neurotransmission. In this perspective, we summarize canonical Cl- transporter expression and investigate altered gene and protein expression of Nkcc1 and Kcc2 in FXS models. We then discuss interactions between Cl- transporters and neurotransmission complexes, and how these links could cause imbalances in inhibitory neurotransmission that may alter mature circuits. Finally, we highlight current therapeutic strategies and promising new directions in targeting Cl- transporter expression in FXS patients.
Collapse
Affiliation(s)
| | - Caleb Andrew Doll
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
3
|
From bench to bedside: The mGluR5 system in people with and without Autism Spectrum Disorder and animal model systems. Transl Psychiatry 2022; 12:395. [PMID: 36127322 PMCID: PMC9489881 DOI: 10.1038/s41398-022-02143-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/19/2022] [Accepted: 09/01/2022] [Indexed: 11/08/2022] Open
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is a key regulator of excitatory (E) glutamate and inhibitory (I) γ-amino butyric acid (GABA) signalling in the brain. Despite the close functional ties between mGluR5 and E/I signalling, no-one has directly examined the relationship between mGluR5 and glutamate or GABA in vivo in the human brain of autistic individuals. We measured [18F] FPEB (18F-3-fluoro-5-[(pyridin-3-yl)ethynyl]benzonitrile) binding in 15 adults (6 with Autism Spectrum Disorder) using two regions of interest, the left dorsomedial prefrontal cortex and a region primarily composed of left striatum and thalamus. These two regions were mapped out using MEGA-PRESS voxels and then superimposed on reconstructed PET images. This allowed for direct comparison between mGluR5, GABA + and Glx. To better understand the molecular underpinnings of our results we used an autoradiography study of mGluR5 in three mouse models associated with ASD: Cntnap2 knockout, Shank3 knockout, and 16p11.2 deletion. Autistic individuals had significantly higher [18F] FPEB binding (t (13) = -2.86, p = 0.047) in the left striatum/thalamus region of interest as compared to controls. Within this region, there was a strong negative correlation between GABA + and mGluR5 density across the entire cohort (Pearson's correlation: r (14) = -0.763, p = 0.002). Cntnap2 KO mice had significantly higher mGlu5 receptor binding in the striatum (caudate-putamen) as compared to wild-type (WT) mice (n = 15, p = 0.03). There were no differences in mGluR5 binding for mice with the Shank3 knockout or 16p11.2 deletion. Given that Cntnap2 is associated with a specific striatal deficit of parvalbumin positive GABA interneurons and 'autistic' features, our findings suggest that an increase in mGluR5 in ASD may relate to GABAergic interneuron abnormalities.
Collapse
|
4
|
Purushotham SS, Reddy NMN, D'Souza MN, Choudhury NR, Ganguly A, Gopalakrishna N, Muddashetty R, Clement JP. A perspective on molecular signalling dysfunction, its clinical relevance and therapeutics in autism spectrum disorder. Exp Brain Res 2022; 240:2525-2567. [PMID: 36063192 DOI: 10.1007/s00221-022-06448-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022]
Abstract
Intellectual disability (ID) and autism spectrum disorder (ASD) are neurodevelopmental disorders that have become a primary clinical and social concern, with a prevalence of 2-3% in the population. Neuronal function and behaviour undergo significant malleability during the critical period of development that is found to be impaired in ID/ASD. Human genome sequencing studies have revealed many genetic variations associated with ASD/ID that are further verified by many approaches, including many mouse and other models. These models have facilitated the identification of fundamental mechanisms underlying the pathogenesis of ASD/ID, and several studies have proposed converging molecular pathways in ASD/ID. However, linking the mechanisms of the pathogenic genes and their molecular characteristics that lead to ID/ASD has progressed slowly, hampering the development of potential therapeutic strategies. This review discusses the possibility of recognising the common molecular causes for most ASD/ID based on studies from the available models that may enable a better therapeutic strategy to treat ID/ASD. We also reviewed the potential biomarkers to detect ASD/ID at early stages that may aid in diagnosis and initiating medical treatment, the concerns with drug failure in clinical trials, and developing therapeutic strategies that can be applied beyond a particular mutation associated with ASD/ID.
Collapse
Affiliation(s)
- Sushmitha S Purushotham
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Neeharika M N Reddy
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Michelle Ninochka D'Souza
- Centre for Brain Research, Indian Institute of Science Campus, CV Raman Avenue, Bangalore, 560 012, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - Nilpawan Roy Choudhury
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Anusa Ganguly
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Niharika Gopalakrishna
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Ravi Muddashetty
- Centre for Brain Research, Indian Institute of Science Campus, CV Raman Avenue, Bangalore, 560 012, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India.
| |
Collapse
|
5
|
Lim S, Lee S. Chemical Modulators for Targeting Autism Spectrum Disorders: From Bench to Clinic. Molecules 2022; 27:molecules27165088. [PMID: 36014340 PMCID: PMC9414776 DOI: 10.3390/molecules27165088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by diverse behavioral symptoms such as repetitive behaviors, social deficits, anxiety, hyperactivity, and irritability. Despite their increasing incidence, the specific pathological mechanisms of ASD are still unknown, and the degree and types of symptoms that vary from patient to patient make it difficult to develop drugs that target the core symptoms of ASD. Although various atypical antipsychotics and antidepressants have been applied to regulate ASD symptoms, these drugs can only alleviate the symptoms and do not target the major causes. Therefore, development of novel drugs targeting factors directly related to the onset of ASD is required. Among the various factors related to the onset of ASD, several chemical modulators to treat ASD, focused on serotonin (5-hydroxytryptamine, 5-HT) and glutamate receptors, microbial metabolites, and inflammatory cytokines, are explored in this study. In particular, we focus on the chemical drugs that have improved various aspects of ASD symptoms in animal models and in clinical trials for various ages of patients with ASD.
Collapse
Affiliation(s)
- Songhyun Lim
- Creative Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Sanghee Lee
- Creative Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
- Department of HY-KIST Bio-Convergence, Hanyang University, Seoul 04763, Korea
- Correspondence: ; Tel.: +82-2-958-5138
| |
Collapse
|
6
|
Jiang CC, Lin LS, Long S, Ke XY, Fukunaga K, Lu YM, Han F. Signalling pathways in autism spectrum disorder: mechanisms and therapeutic implications. Signal Transduct Target Ther 2022; 7:229. [PMID: 35817793 PMCID: PMC9273593 DOI: 10.1038/s41392-022-01081-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a prevalent and complex neurodevelopmental disorder which has strong genetic basis. Despite the rapidly rising incidence of autism, little is known about its aetiology, risk factors, and disease progression. There are currently neither validated biomarkers for diagnostic screening nor specific medication for autism. Over the last two decades, there have been remarkable advances in genetics, with hundreds of genes identified and validated as being associated with a high risk for autism. The convergence of neuroscience methods is becoming more widely recognized for its significance in elucidating the pathological mechanisms of autism. Efforts have been devoted to exploring the behavioural functions, key pathological mechanisms and potential treatments of autism. Here, as we highlight in this review, emerging evidence shows that signal transduction molecular events are involved in pathological processes such as transcription, translation, synaptic transmission, epigenetics and immunoinflammatory responses. This involvement has important implications for the discovery of precise molecular targets for autism. Moreover, we review recent insights into the mechanisms and clinical implications of signal transduction in autism from molecular, cellular, neural circuit, and neurobehavioural aspects. Finally, the challenges and future perspectives are discussed with regard to novel strategies predicated on the biological features of autism.
Collapse
Affiliation(s)
- Chen-Chen Jiang
- International Joint Laboratory for Drug Target of Critical Illnesses; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Li-Shan Lin
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Sen Long
- Department of Pharmacy, Hangzhou Seventh People's Hospital, Mental Health Center Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Xiao-Yan Ke
- Child Mental Health Research Center, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Ying-Mei Lu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Feng Han
- International Joint Laboratory for Drug Target of Critical Illnesses; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
7
|
Davidson M, Sebastian SA, Benitez Y, Desai S, Quinonez J, Ruxmohan S, Stein JD, Cueva W. Behavioral Problems in Fragile X Syndrome: A Review of Clinical Management. Cureus 2022; 14:e21840. [PMID: 35291526 PMCID: PMC8896844 DOI: 10.7759/cureus.21840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 01/08/2023] Open
Abstract
Fragile X syndrome (FXS) is noted to be the leading cause of inherited intellectual disabilities and is caused by expansive cytosine-guanine-guanine (CGG) trinucleotide repeats in the fragile X mental retardation 1 gene (FMR1). FXS can display a wide range of behavioral problems in addition to intellectual and developmental issues. Management of these problems includes both pharmacological and non-pharmacological options and research on these different management styles has been extensive in recent years. This narrative review aimed to collate recent evidence on the various management options of behavioral problems in FXS, including the pharmacological and non-pharmacological treatments, and also to provide a review of the newer avenues in the FXS treatment.
Collapse
Affiliation(s)
| | | | | | - Shreeya Desai
- Research, Larkin Community Hospital, South Miami, USA
| | - Jonathan Quinonez
- Neurology/Osteopathic Neuromuscular Medicine, Larkin Community Hospital, South Miami, USA
| | | | - Joel D Stein
- Osteopathic Neuromuscular Medicine, Family Medicine, Sports Medicine, Pain Medicine, Lake Erie College of Osteopathic Medicine (LECOM) Bradenton, Bradenton, USA.,Pain Management, Osteopathic Neuromuscular Medicine, Sports Medicine, Larkin Community Hospital, South Miami, USA
| | - Wilson Cueva
- Neurology, Larkin Community Hospital, South Miami, USA
| |
Collapse
|
8
|
Vallés AS, Barrantes FJ. Dendritic spine membrane proteome and its alterations in autistic spectrum disorder. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:435-474. [PMID: 35034726 DOI: 10.1016/bs.apcsb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic spines are small protrusions stemming from the dendritic shaft that constitute the primary specialization for receiving and processing excitatory neurotransmission in brain synapses. The disruption of dendritic spine function in several neurological and neuropsychiatric diseases leads to severe information-processing deficits with impairments in neuronal connectivity and plasticity. Spine dysregulation is usually accompanied by morphological alterations to spine shape, size and/or number that may occur at early pathophysiological stages and not necessarily be reflected in clinical manifestations. Autism spectrum disorder (ASD) is one such group of diseases involving changes in neuronal connectivity and abnormal morphology of dendritic spines on postsynaptic neurons. These alterations at the subcellular level correlate with molecular changes in the spine proteome, with alterations in the copy number, topography, or in severe cases in the phenotype of the molecular components, predominantly of those proteins involved in spine recognition and adhesion, reflected in abnormally short lifetimes of the synapse and compensatory increases in synaptic connections. Since cholinergic neurotransmission participates in the regulation of cognitive function (attention, memory, learning processes, cognitive flexibility, social interactions) brain acetylcholine receptors are likely to play an important role in the dysfunctional synapses in ASD, either directly or indirectly via the modulatory functions exerted on other neurotransmitter receptor proteins and spine-resident proteins.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
9
|
|
10
|
Soorya LV, Fogg L, Ocampo E, Printen M, Youngkin S, Halpern D, Kolevzon A, Lee S, Grodberg D, Anagnostou E. Neurocognitive Outcomes from Memantine: A Pilot, Double-Blind, Placebo-Controlled Trial in Children with Autism Spectrum Disorder. J Child Adolesc Psychopharmacol 2021; 31:475-484. [PMID: 34543081 DOI: 10.1089/cap.2021.0010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: Studies interrogating therapeutics which alter the excitation-inhibition balance in the treatment of autism spectrum disorder (ASD) have reported mixed results on social and behavioral outcomes. Methods: The aim of this randomized, double-blind placebo-controlled pilot trial was to evaluate neurocognitive effects of memantine over a 24-week trial. Twenty-three children ages 6-12 years old with ASD were randomized to memantine or placebo. Primary outcomes included measures of apraxia and expressive language with evaluations at midpoint (week 12) and endpoint (week 24). Secondary outcomes included memory and adaptive behavior measures. Exploratory outcomes included changes in overall cognitive functioning and behavior (e.g., Aberrant Behavior Checklist). Results: Results suggest that memantine was well-tolerated. Dropout rates were high across groups with only 14 participants completing the 6-month trial. Memantine was not associated with improvements in apraxia and expressive language. Treatment with memantine was associated with improvements in verbal recognition memory as measured by the Narrative Memory-Recognition (NEPSY-II) (F = 5.05, p = .03). In addition, exploratory analyses of changes in Intelligence quotient (IQ) suggest improvements on verbal IQ (d = 1.8). Conclusions: Results suggest future studies of memantine in ASD may benefit from shifting treatment targets from social and behavioral outcomes to exploration of effects of memantine on cognition, potentially as an adjunct to learning and educational interventions. ClinicalTrials.gov: NCT01372449.
Collapse
Affiliation(s)
- Latha Valluripalli Soorya
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Louis Fogg
- Department of Nursing, Rush University Medical Center, Chicago, Illinois, USA
| | - Edith Ocampo
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madison Printen
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sarah Youngkin
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Danielle Halpern
- Department of Psychiatry, Icahn School of Medicine, New York, New York, USA
| | - Alexander Kolevzon
- Department of Psychiatry, Icahn School of Medicine, New York, New York, USA
| | - Soo Lee
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - David Grodberg
- Child Study Center, Yale University, New Haven, Connecticut, USA
| | | |
Collapse
|
11
|
The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules 2021; 26:molecules26164913. [PMID: 34443494 PMCID: PMC8400259 DOI: 10.3390/molecules26164913] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Taurine is a naturally occurring sulfur-containing amino acid that is found abundantly in excitatory tissues, such as the heart, brain, retina and skeletal muscles. Taurine was first isolated in the 1800s, but not much was known about this molecule until the 1990s. In 1985, taurine was first approved as the treatment among heart failure patients in Japan. Accumulating studies have shown that taurine supplementation also protects against pathologies associated with mitochondrial defects, such as aging, mitochondrial diseases, metabolic syndrome, cancer, cardiovascular diseases and neurological disorders. In this review, we will provide a general overview on the mitochondria biology and the consequence of mitochondrial defects in pathologies. Then, we will discuss the antioxidant action of taurine, particularly in relation to the maintenance of mitochondria function. We will also describe several reported studies on the current use of taurine supplementation in several mitochondria-associated pathologies in humans.
Collapse
|
12
|
Salcedo-Arellano MJ, Cabal-Herrera AM, Punatar RH, Clark CJ, Romney CA, Hagerman RJ. Overlapping Molecular Pathways Leading to Autism Spectrum Disorders, Fragile X Syndrome, and Targeted Treatments. Neurotherapeutics 2021; 18:265-283. [PMID: 33215285 PMCID: PMC8116395 DOI: 10.1007/s13311-020-00968-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) are subdivided into idiopathic (unknown) etiology and secondary, based on known etiology. There are hundreds of causes of ASD and most of them are genetic in origin or related to the interplay of genetic etiology and environmental toxicology. Approximately 30 to 50% of the etiologies can be identified when using a combination of available genetic testing. Many of these gene mutations are either core components of the Wnt signaling pathway or their modulators. The full mutation of the fragile X mental retardation 1 (FMR1) gene leads to fragile X syndrome (FXS), the most common cause of monogenic origin of ASD, accounting for ~ 2% of the cases. There is an overlap of molecular mechanisms in those with idiopathic ASD and those with FXS, an interaction between various signaling pathways is suggested during the development of the autistic brain. This review summarizes the cross talk between neurobiological pathways found in ASD and FXS. These signaling pathways are currently under evaluation to target specific treatments in search of the reversal of the molecular abnormalities found in both idiopathic ASD and FXS.
Collapse
Affiliation(s)
- Maria Jimena Salcedo-Arellano
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| | - Ana Maria Cabal-Herrera
- Group on Congenital Malformations and Dysmorphology, Faculty of Health, Universidad del Valle, Cali, 00000, Colombia
| | - Ruchi Harendra Punatar
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Courtney Jessica Clark
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Christopher Allen Romney
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Randi J Hagerman
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| |
Collapse
|
13
|
Glutamatergic Dysfunction and Synaptic Ultrastructural Alterations in Schizophrenia and Autism Spectrum Disorder: Evidence from Human and Rodent Studies. Int J Mol Sci 2020; 22:ijms22010059. [PMID: 33374598 PMCID: PMC7793137 DOI: 10.3390/ijms22010059] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The correlation between dysfunction in the glutamatergic system and neuropsychiatric disorders, including schizophrenia and autism spectrum disorder, is undisputed. Both disorders are associated with molecular and ultrastructural alterations that affect synaptic plasticity and thus the molecular and physiological basis of learning and memory. Altered synaptic plasticity, accompanied by changes in protein synthesis and trafficking of postsynaptic proteins, as well as structural modifications of excitatory synapses, are critically involved in the postnatal development of the mammalian nervous system. In this review, we summarize glutamatergic alterations and ultrastructural changes in synapses in schizophrenia and autism spectrum disorder of genetic or drug-related origin, and briefly comment on the possible reversibility of these neuropsychiatric disorders in the light of findings in regular synaptic physiology.
Collapse
|
14
|
Recent Advances in the Pharmacological Management of Behavioral Disturbances Associated with Autism Spectrum Disorder in Children and Adolescents. Paediatr Drugs 2020; 22:473-483. [PMID: 32686015 DOI: 10.1007/s40272-020-00408-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neuropsychiatric condition affecting an estimated one in 36 children. Youth with ASD may have severe behavioral disturbances including irritability, aggression, and hyperactivity. Currently, there are only two medications (risperidone and aripiprazole) approved by the US Food and Drug Administration (FDA) for the treatment of irritability associated with ASD. Pharmacologic treatments are commonly used to target ASD-associated symptoms including irritability, mood lability, anxiety, and hyperactivity. However, evidence for the efficacy of many commonly used treatments is limited by the lack of large placebo-controlled trials of these medications in this population. Research into the pathophysiology of ASD has led to new targets for pharmacologic therapy including the neuroimmune system, the endocannabinoid system, and the glutamatergic neurotransmitter system. The goal of this review is to provide an overview of the current evidence base for commonly used treatments, as well as emerging treatment options for common behavioral disturbances seen in youth with ASD.
Collapse
|
15
|
Sharma R, Rahi S, Mehan S. Neuroprotective potential of solanesol in intracerebroventricular propionic acid induced experimental model of autism: Insights from behavioral and biochemical evidence. Toxicol Rep 2019; 6:1164-1175. [PMID: 31763180 PMCID: PMC6861559 DOI: 10.1016/j.toxrep.2019.10.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
Autism is the category used within the newest edition of the diagnostic and statistical manual of neurodevelopmental disorders. Autism is a spectrum of disorder where a variety of behavioural patterns observed in autistic patients, such as stereotypes and repetitive behavior, hyperexcitability, depression-like symptoms, and memory and cognitive dysfunctions. Neuropathological hallmarks that associated with autism are mitochondrial dysfunction, oxidative stress, neuroinflammation, Neuro-excitation, abnormal synapse formation, overexpression of glial cells in specific brain regions like cerebellum, cerebral cortex, amygdala, and hippocampus. ICV injection of propionic acid (PPA) (4 μl/0.26 M) mimics autistic-like behavioral and biochemical alterations in rats. Literature findings reveal that there is a link between autism neuronal mitochondrial coenzyme-Q10 (CoQ10) and ETC-complexes dysfunctions are the keys pathogenic events for autism. Therefore, in the current study, we explore the neuroprotective interventions of Solanesol (SNL) 40 and 60 mg/kg alone and in combination with standard drugs Aripiprazole (ARP) 5 mg/kg, Citalopram (CTP) 10 mg/kg, Memantine (MEM) 5 mg/kg and Donepezil (DNP) 3 mg/kg to overcome behavioral and biochemical alterations in PPA induced experimental model of Autism. Chronic treatment with SNL 60 mg/kg in combination with standard drug shows a marked improvement in locomotion, muscle coordination, long-term memory and the decrease in depressive behavior. While, chronic treatment of SNL alone and in combination with standard drug aripiprazole, citalopram, donepezil, and memantine shows the Neuroprotective potential by enhancing the cognitive deficits, biochemical alterations along with reducing the level of inflammatory mediators and oxidative stress.
Collapse
Key Words
- AChE, acetylcholinesterase acetylcholinesterase
- ARP, Aripiprazole
- ATP
- Aripiprazole
- Autism
- BBB, blood-brain barrier
- CNS, center nerves system
- CTP, Citalopram
- Citalopram
- CoQ10, coenzyme-Q10
- Coenzyme-Q10
- DNP, Donepezil
- Donepezil
- ELT, escape latency
- ETC, electron-transport chain
- ICV, Intracerebroventricular
- LDH, lactate dehydrogenase
- MAPK3, mitogen-activated protein kinase 3
- MDA, malondialdehyde
- MEM, Memantine
- Memantine
- NO, nitric oxide
- PPA, propionic acid
- Propionic acid
- SNL, Solanesol
- SOD, superoxide dismutase
- UBE3A, Ubiquitin-protein ligase E3A
- i.p., Intraperitoneal route
- mitochondrial dysfunction
- p.o., Oral
Collapse
Affiliation(s)
| | | | - Sidharth Mehan
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
16
|
Hong MP, Erickson CA. Investigational drugs in early-stage clinical trials for autism spectrum disorder. Expert Opin Investig Drugs 2019; 28:709-718. [PMID: 31352835 DOI: 10.1080/13543784.2019.1649656] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Pharmacologic interventions in Autism Spectrum Disorder (ASD) have historically focused on symptom-based approaches. However, a treatment for the core social deficits has remained unidentified. While a definitive theory for the cause of ASD is not yet known, recent advances in our understanding of ASD pathophysiology have opened the door for research on new pharmaceutical methods to target core symptomology. Areas covered: Herein, we review the novel pharmacologic therapies undergoing early-stage clinical trials for the treatment of the social symptoms associated with ASD. Specifically, these strategies center on altering neurologic excitatory and inhibitory imbalance, neuropeptide abnormalities, immunologic dysfunction, and biochemical deficiencies in ASD. Expert opinion: Utilizing the growing field of knowledge regarding the pathological mechanisms and altered neurobiology of individuals with ASD has led to the development of many innovative pharmaceutical interventions. Clinical trials for neurobiologic and immunologic targets show promise in impacting the social behavior and processing deficits in ASD but need evaluation in larger clinical trials and continued biomarker development to more effectively and consistently assess pharmacologic effects. Additionally, evaluating patient-specific drug responsivity and integrating behavioral intervention in conjunction with pharmacologic treatment is crucial to developing a successful approach to ASD treatment.
Collapse
Affiliation(s)
- Michael P Hong
- a Division of Psychiatry, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b College of Medicine, University of Cincinnati , Cincinnati , Oh , USA
| | - Craig A Erickson
- a Division of Psychiatry, Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA.,b College of Medicine, University of Cincinnati , Cincinnati , Oh , USA
| |
Collapse
|
17
|
Protic D, Salcedo-Arellano MJ, Dy JB, Potter LA, Hagerman RJ. New Targeted Treatments for Fragile X Syndrome. Curr Pediatr Rev 2019; 15:251-258. [PMID: 31241016 PMCID: PMC6930353 DOI: 10.2174/1573396315666190625110748] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/14/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Fragile X Syndrome (FXS) is the most common cause of inherited intellectual disability with prevalence rates estimated to be 1:5,000 in males and 1:8,000 in females. The increase of >200 Cytosine Guanine Guanine (CGG) repeats in the 5' untranslated region of the Fragile X Mental Retardation 1 (FMR1) gene results in transcriptional silencing on the FMR1 gene with a subsequent reduction or absence of fragile X mental retardation protein (FMRP), an RNA binding protein involved in the maturation and elimination of synapses. In addition to intellectual disability, common features of FXS are behavioral problems, autism, language deficits and atypical physical features. There are still no currently approved curative therapies for FXS, and clinical management continues to focus on symptomatic treatment of comorbid behaviors and psychiatric problems. Here we discuss several treatments that target the neurobiological pathway abnormal in FXS. These medications are clinically available at present and the data suggest that these medications can be helpful for those with FXS.
Collapse
Affiliation(s)
- Dragana Protic
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, United States.,Department of Pharmacology, Clinical Pharmacology and Toxicology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Maria J Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, United States.,Department of Pediatrics, Davis School of Medicine, University of California, Sacramento, CA, United States
| | - Jeanne Barbara Dy
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, United States.,MedMom Institute for Human Development, Pasig City, Philippines.,Department of Pediatrics, The Medical City, Ortigas Avenue, Pasig City, NCR, Philippines.,School of Medicine and Public Health, Ateneo de Manila University, Pasig City, NCR, Philippines
| | - Laura A Potter
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, United States
| | - Randi J Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, United States.,Department of Pediatrics, Davis School of Medicine, University of California, Sacramento, CA, United States
| |
Collapse
|
18
|
Park E, Cohen I, Gonzalez M, Castellano MR, Flory M, Jenkins EC, Brown WT, Schuller-Levis G. Is Taurine a Biomarker in Autistic Spectrum Disorder? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:3-16. [PMID: 28849439 DOI: 10.1007/978-94-024-1079-2_1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Taurine is a sulfur-containing amino acid which is not incorporated into protein. However, taurine has various critical physiological functions including development of the eye and brain, reproduction, osmoregulation, and immune functions including anti-inflammatory as well as anti-oxidant activity. The causes of autistic spectrum disorder (ASD) are not clear but a high heritability implicates an important role for genetic factors. Reports also implicate oxidative stress and inflammation in the etiology of ASD. Thus, taurine, a well-known antioxidant and regulator of inflammation, was investigated here using the sera from both girls and boys with ASD as well as their siblings and parents. Previous reports regarding taurine serum concentrations in ASD from various laboratories have been controversial. To address the potential role of taurine in ASD, we collected sera from 66 children with ASD (males: 45; females: 21, age 1.5-11.5 years, average age 5.2 ± 1.6) as well as their unaffected siblings (brothers: 24; sisters: 32, age 1.5-17 years, average age 7.0 ± 2.0) as controls of the children with ASD along with parents (fathers: 49; mothers: 54, age 28-45 years). The sera from normal adult controls (males: 47; females: 51, age 28-48 years) were used as controls for the parents. Taurine concentrations in all sera samples were measured using high performance liquid chromatography (HPLC) using a phenylisothiocyanate labeling technique. Taurine concentrations from female and male children with ASD were 123.8 ± 15.2 and 145.8 ± 8.1 μM, respectively, and those from their unaffected brothers and sisters were 142.6 ± 10.4 and 150.8 ± 8.4 μM, respectively. There was no significant difference in taurine concentration between autistic children and their unaffected siblings. Taurine concentrations in children with ASD were also not significantly different from their parents (mothers: 139.6 ± 7.7 μM, fathers: 147.4 ± 7.5 μM). No significant difference was observed between adult controls and parents of ASD children (control females: 164.8 ± 4.8 μM, control males: 163.0 ± 7.0 μM). However, 21 out of 66 children with ASD had low taurine concentrations (<106 μM). Since taurine has anti-oxidant activity, children with ASD with low taurine concentrations will be examined for abnormal mitochondrial function. Our data imply that taurine may be a valid biomarker in a subgroup of ASD.
Collapse
Affiliation(s)
- Eunkyue Park
- Departments of Developmental Neurobiology, NY State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Rd, Staten Island, NY, 10314, USA.
| | - Ira Cohen
- Department of Psychology, NY State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Maripaz Gonzalez
- Department of Psychology, NY State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Mario R Castellano
- Division of Hematology/Oncology, Division of Research, Department of Medicine, Staten Island University Hospital-Northwell Health, Staten Island, NY, 10305, USA
| | - Michael Flory
- Department of Infant Development, NY State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Edmund C Jenkins
- Department of Human Genetics, NY State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - W Ted Brown
- Department of Human Genetics, NY State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Georgia Schuller-Levis
- Departments of Developmental Neurobiology, NY State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Rd, Staten Island, NY, 10314, USA
| |
Collapse
|
19
|
Lamy M, Erickson CA. Pharmacological management of behavioral disturbances in children and adolescents with autism spectrum disorders. Curr Probl Pediatr Adolesc Health Care 2018; 48:250-264. [PMID: 30262163 DOI: 10.1016/j.cppeds.2018.08.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neuropsychiatric condition that, based on recent CDC estimates affects an estimated 1 in 59 American children. Behavioral treatments remain the mainstay of treatment for the core symptoms of ASD including communication deficits, social interaction deficits and repetitive behavior. However, youth with ASD may also have severe behavioral challenges including irritability, aggression, and hyperactivity. Currently there are only two medications (risperidone and aripiprazole) approved by the FDA for the treatment of irritability associated with ASD in children. Psychiatric comorbidities are common in youth with ASD, affecting up to 70% of affected children and adolescents. Given the burden of co-occurring disorders, medications are often employed to target symptoms such as irritability, anxiety, and hyperactivity. Other common co-occurring conditions including gastrointestinal disorders and sleep disorders may be improved with pharmacologic management. Evidence for the efficacy of many commonly used psychotropic medications in ASD is limited by the lack of large placebo-controlled trials in youth with ASD. This paper reviews the current literature regarding use of medications to address co-occurring conditions in children and adolescents with ASD as well as areas of emerging research.
Collapse
Affiliation(s)
- Martine Lamy
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States.
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| |
Collapse
|
20
|
Green J, Garg S. Annual Research Review: The state of autism intervention science: progress, target psychological and biological mechanisms and future prospects. J Child Psychol Psychiatry 2018; 59:424-443. [PMID: 29574740 DOI: 10.1111/jcpp.12892] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND There has been recent systematic review of key evidence in psychosocial intervention in autism but little review of biological treatments. METHODS We analyse the current literature from the perspective of intervention and mechanism targets across social and biological development. RESULTS The overall quality of trials evidence in autism intervention remains relatively low, despite some recent progress. Many treatments in common use have little or no evidence base. This is very concerning in such an important disorder. A variety of psychosocial interventions can show effect to improve some short-term effects on children's immediate dyadic social interactions, for instance with caregivers. But showing true effectiveness in this developmental disorder requires generalisation of such effects into wider social contexts, on autism symptoms and in long-term progress in development. Only a few interventions so far have begun to show this. A number of early phase interventions on biological targets have shown real promise, but none has yet progressed to larger scale effectiveness trials on behavioural or symptom outcomes. CONCLUSIONS There has been enough progress in psychosocial intervention research now to be able to begin to identify some evidence-based practice in autism treatment. To consolidate and improve outcomes, the next phase of intervention research needs improved trial design, and an iterative approach building on success. It may also include the testing of potential synergies between promising biological and psychosocial interventions.
Collapse
Affiliation(s)
- Jonathan Green
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Manchester University NHS Foundation Trust, Manchester, UK.,Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Shruti Garg
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Manchester University NHS Foundation Trust, Manchester, UK.,Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| |
Collapse
|
21
|
Hutson RL, Thompson RL, Bantel AP, Tessier CR. Acamprosate rescues neuronal defects in the Drosophila model of Fragile X Syndrome. Life Sci 2018; 195:65-70. [PMID: 29317220 DOI: 10.1016/j.lfs.2018.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 10/18/2022]
Abstract
AIMS Several off-label studies have shown that acamprosate can provide some clinical benefits in youth with Fragile X Syndrome (FXS), an autism spectrum disorder caused by loss of function of the highly conserved FMR1 gene. This study investigated the ability of acamprosate to rescue cellular, molecular and behavioral defects in the Drosophila model of FXS. MAIN METHODS A high (100μM) and low (10μM) dose of acamprosate was fed to Drosophila FXS (dfmr1 null) or genetic control (w1118) larvae and then analyzed in multiple paradigms. A larval crawling assay was used to monitor aberrant FXS behavior, overgrowth of the neuromuscular junction (NMJ) was quantified to assess neuronal development, and quantitative RT-PCR was used to evaluate expression of deregulated cbp53E mRNA. KEY FINDINGS Acamprosate treatment partially or completely rescued all of the FXS phenotypes analyzed, according to dose. High doses rescued cellular overgrowth and dysregulated cbp53E mRNA expression, but aberrant crawling behavior was not affected. Low doses of acamprosate, however, did not affect synapse number at the NMJ, but could rescue NMJ overgrowth, locomotor defects, and cbp53E mRNA expression. This dual nature of acamprosate suggests multiple molecular mechanisms may be involved in acamprosate function depending on the dosage used. SIGNIFICANCE Acamprosate may be a useful therapy for FXS and potentially other autism spectrum disorders. However, understanding the molecular mechanisms involved with different doses of this drug will likely be necessary to obtain optimal results.
Collapse
Affiliation(s)
- Russell L Hutson
- Department of Biological Sciences, University of Notre Dame, South Bend, IN, United States
| | - Rachel L Thompson
- Department of Biological Sciences, University of Notre Dame, South Bend, IN, United States
| | - Andrew P Bantel
- Department of Medical and Molecular Genetics, Indiana University School of Medicine-South Bend, South Bend, IN, United States
| | - Charles R Tessier
- Department of Medical and Molecular Genetics, Indiana University School of Medicine-South Bend, South Bend, IN, United States.
| |
Collapse
|
22
|
McClellan L, Dominick KC, Pedapati EV, Wink LK, Erickson CA. Lurasidone for the treatment of irritability and anger in autism spectrum disorders. Expert Opin Investig Drugs 2017; 26:985-989. [DOI: 10.1080/13543784.2017.1353600] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lynn McClellan
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kelli C. Dominick
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ernest V. Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Logan K. Wink
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Craig A. Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
23
|
Singh R, Turner RC, Nguyen L, Motwani K, Swatek M, Lucke-Wold BP. Pediatric Traumatic Brain Injury and Autism: Elucidating Shared Mechanisms. Behav Neurol 2016; 2016:8781725. [PMID: 28074078 PMCID: PMC5198096 DOI: 10.1155/2016/8781725] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
Pediatric traumatic brain injury (TBI) and autism spectrum disorder (ASD) are two serious conditions that affect youth. Recent data, both preclinical and clinical, show that pediatric TBI and ASD share not only similar symptoms but also some of the same biologic mechanisms that cause these symptoms. Prominent symptoms for both disorders include gastrointestinal problems, learning difficulties, seizures, and sensory processing disruption. In this review, we highlight some of these shared mechanisms in order to discuss potential treatment options that might be applied for each condition. We discuss potential therapeutic and pharmacologic options as well as potential novel drug targets. Furthermore, we highlight advances in understanding of brain circuitry that is being propelled by improved imaging modalities. Going forward, advanced imaging will help in diagnosis and treatment planning strategies for pediatric patients. Lessons from each field can be applied to design better and more rigorous trials that can be used to improve guidelines for pediatric patients suffering from TBI or ASD.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Linda Nguyen
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Kartik Motwani
- Department of Medical Sciences, University of Florida School of Medicine, Gainesville, FL 32611, USA
| | - Michelle Swatek
- Department of Psychology, North Carolina State University, Raleigh, NC 27695, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| |
Collapse
|
24
|
Brondino N, Fusar-Poli L, Panisi C, Damiani S, Barale F, Politi P. Pharmacological Modulation of GABA Function in Autism Spectrum Disorders: A Systematic Review of Human Studies. J Autism Dev Disord 2016; 46:825-39. [PMID: 26443675 DOI: 10.1007/s10803-015-2619-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autism spectrum disorders are an emerging health problem worldwide, but little is known about their pathogenesis. It has been hypothesized that autism may result from an imbalance between excitatory glutamatergic and inhibitory GABAergic pathways. Commonly used medications such as valproate, acamprosate, and arbaclofen may act on the GABAergic system and be a potential treatment for people with ASD. The present systematic review aimed at evaluating the state-of-the-art of clinical trials of GABA modulators in autism. To date there is insufficient evidence to suggest the use of these drugs in autistic subjects, even if data are promising. Of note, short-term use of all the reviewed medications appears to be safe. Future well designed trials are needed to elucidate these preliminary findings.
Collapse
Affiliation(s)
- Natascia Brondino
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy.
| | - Laura Fusar-Poli
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Cristina Panisi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Stefano Damiani
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Francesco Barale
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| |
Collapse
|
25
|
Choi CH, Schoenfeld BP, Bell AJ, Hinchey J, Rosenfelt C, Gertner MJ, Campbell SR, Emerson D, Hinchey P, Kollaros M, Ferrick NJ, Chambers DB, Langer S, Sust S, Malik A, Terlizzi AM, Liebelt DA, Ferreiro D, Sharma A, Koenigsberg E, Choi RJ, Louneva N, Arnold SE, Featherstone RE, Siegel SJ, Zukin RS, McDonald TV, Bolduc FV, Jongens TA, McBride SMJ. Multiple Drug Treatments That Increase cAMP Signaling Restore Long-Term Memory and Aberrant Signaling in Fragile X Syndrome Models. Front Behav Neurosci 2016; 10:136. [PMID: 27445731 PMCID: PMC4928101 DOI: 10.3389/fnbeh.2016.00136] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/15/2016] [Indexed: 01/01/2023] Open
Abstract
Fragile X is the most common monogenic disorder associated with intellectual disability (ID) and autism spectrum disorders (ASD). Additionally, many patients are afflicted with executive dysfunction, ADHD, seizure disorder and sleep disturbances. Fragile X is caused by loss of FMRP expression, which is encoded by the FMR1 gene. Both the fly and mouse models of fragile X are also based on having no functional protein expression of their respective FMR1 homologs. The fly model displays well defined cognitive impairments and structural brain defects and the mouse model, although having subtle behavioral defects, has robust electrophysiological phenotypes and provides a tool to do extensive biochemical analysis of select brain regions. Decreased cAMP signaling has been observed in samples from the fly and mouse models of fragile X as well as in samples derived from human patients. Indeed, we have previously demonstrated that strategies that increase cAMP signaling can rescue short term memory in the fly model and restore DHPG induced mGluR mediated long term depression (LTD) in the hippocampus to proper levels in the mouse model (McBride et al., 2005; Choi et al., 2011, 2015). Here, we demonstrate that the same three strategies used previously with the potential to be used clinically, lithium treatment, PDE-4 inhibitor treatment or mGluR antagonist treatment can rescue long term memory in the fly model and alter the cAMP signaling pathway in the hippocampus of the mouse model.
Collapse
Affiliation(s)
- Catherine H Choi
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva UniversityBronx, NY, USA; Department of Dermatology, Dermatology Clinic, Drexel University College of MedicinePhiladelphia, PA, USA; Jongens Laboratory, Department of Genetics, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| | - Brian P Schoenfeld
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva UniversityBronx, NY, USA; Jongens Laboratory, Department of Genetics, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| | - Aaron J Bell
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva UniversityBronx, NY, USA; Jongens Laboratory, Department of Genetics, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| | - Joseph Hinchey
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Cory Rosenfelt
- Bolduc Laboratory, Department of Pediatrics, Center for Neuroscience, University of Alberta Edmonton, AB, Canada
| | - Michael J Gertner
- Zukin Laboratory, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Sean R Campbell
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Danielle Emerson
- Jongens Laboratory, Department of Genetics, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Paul Hinchey
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Maria Kollaros
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Neal J Ferrick
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva UniversityBronx, NY, USA; Jongens Laboratory, Department of Genetics, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| | - Daniel B Chambers
- Bolduc Laboratory, Department of Pediatrics, Center for Neuroscience, University of Alberta Edmonton, AB, Canada
| | - Steven Langer
- Bolduc Laboratory, Department of Pediatrics, Center for Neuroscience, University of Alberta Edmonton, AB, Canada
| | - Steven Sust
- Siegel Laboratory, Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Aatika Malik
- Jongens Laboratory, Department of Genetics, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Allison M Terlizzi
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - David A Liebelt
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - David Ferreiro
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Ali Sharma
- Zukin Laboratory, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Eric Koenigsberg
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Richard J Choi
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Natalia Louneva
- Arnold Laboratory, Department of Psychiatry, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Steven E Arnold
- Arnold Laboratory, Department of Psychiatry, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Robert E Featherstone
- Siegel Laboratory, Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Steven J Siegel
- Siegel Laboratory, Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - R Suzanne Zukin
- Zukin Laboratory, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Thomas V McDonald
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva University Bronx, NY, USA
| | - Francois V Bolduc
- Bolduc Laboratory, Department of Pediatrics, Center for Neuroscience, University of Alberta Edmonton, AB, Canada
| | - Thomas A Jongens
- Jongens Laboratory, Department of Genetics, University of Pennsylvania School of Medicine Philadelphia, PA, USA
| | - Sean M J McBride
- McDonald Laboratory, Section of Molecular Cardiology, Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Yeshiva UniversityBronx, NY, USA; Jongens Laboratory, Department of Genetics, University of Pennsylvania School of MedicinePhiladelphia, PA, USA; Siegel Laboratory, Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania School of MedicinePhiladelphia, PA, USA
| |
Collapse
|
26
|
Frye RE, Rossignol DA. Identification and Treatment of Pathophysiological Comorbidities of Autism Spectrum Disorder to Achieve Optimal Outcomes. CLINICAL MEDICINE INSIGHTS-PEDIATRICS 2016; 10:43-56. [PMID: 27330338 PMCID: PMC4910649 DOI: 10.4137/cmped.s38337] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/15/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023]
Abstract
Despite the fact that the prevalence of autism spectrum disorder (ASD) continues to rise, no effective medical treatments have become standard of care. In this paper we review some of the pathophysiological abnormalities associated with ASD and their potential associated treatments. Overall, there is evidence for some children with ASD being affected by seizure and epilepsy, neurotransmitter dysfunction, sleep disorders, metabolic abnormalities, including abnormalities in folate, cobalamin, tetrahydrobiopterin, carnitine, redox and mitochondrial metabolism, and immune and gastrointestinal disorders. Although evidence for an association between these pathophysiological abnormalities and ASD exists, the exact relationship to the etiology of ASD and its associated symptoms remains to be further defined in many cases. Despite these limitations, treatments targeting some of these pathophysiological abnormalities have been studied in some cases with high-quality studies, whereas treatments for other pathophysiological abnormalities have not been well studied in many cases. There are some areas of more promising treatments specific for ASD including neurotransmitter abnormalities, particularly imbalances in glutamate and acetylcholine, sleep onset disorder (with behavioral therapy and melatonin), and metabolic abnormalities in folate, cobalamin, tetrahydrobiopterin, carnitine, and redox pathways. There is some evidence for treatments of epilepsy and seizures, mitochondrial and immune disorders, and gastrointestinal abnormalities, particularly imbalances in the enteric microbiome, but further clinical studies are needed in these areas to better define treatments specific to children with ASD. Clearly, there are some promising areas of ASD research that could lead to novel treatments that could become standard of care in the future, but more research is needed to better define subgroups of children with ASD who are affected by specific pathophysiological abnormalities and the optimal treatments for these abnormalities.
Collapse
Affiliation(s)
- Richard E Frye
- Arkansas Children's Research Institute, Little Rock, AR, USA.; Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | |
Collapse
|
27
|
Kazdoba TM, Leach PT, Yang M, Silverman JL, Solomon M, Crawley JN. Translational Mouse Models of Autism: Advancing Toward Pharmacological Therapeutics. Curr Top Behav Neurosci 2016; 28:1-52. [PMID: 27305922 PMCID: PMC5116923 DOI: 10.1007/7854_2015_5003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal models provide preclinical tools to investigate the causal role of genetic mutations and environmental factors in the etiology of autism spectrum disorder (ASD). Knockout and humanized knock-in mice, and more recently knockout rats, have been generated for many of the de novo single gene mutations and copy number variants (CNVs) detected in ASD and comorbid neurodevelopmental disorders. Mouse models incorporating genetic and environmental manipulations have been employed for preclinical testing of hypothesis-driven pharmacological targets, to begin to develop treatments for the diagnostic and associated symptoms of autism. In this review, we summarize rodent behavioral assays relevant to the core features of autism, preclinical and clinical evaluations of pharmacological interventions, and strategies to improve the translational value of rodent models of autism.
Collapse
Affiliation(s)
- Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Prescott T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Mu Yang
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Marjorie Solomon
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
28
|
Terbeck S, Akkus F, Chesterman LP, Hasler G. The role of metabotropic glutamate receptor 5 in the pathogenesis of mood disorders and addiction: combining preclinical evidence with human Positron Emission Tomography (PET) studies. Front Neurosci 2015; 9:86. [PMID: 25852460 PMCID: PMC4364244 DOI: 10.3389/fnins.2015.00086] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/27/2015] [Indexed: 12/30/2022] Open
Abstract
In the present review, we deliver an overview of the involvement of metabotropic glutamate receptor 5 (mGluR5) activity and density in pathological anxiety, mood disorders and addiction. Specifically, we will describe mGluR5 studies in humans that employed Positron Emission Tomography (PET) and combined the findings with preclinical animal research. This combined view of different methodological approaches—from basic neurobiological approaches to human studies—might give a more comprehensive and clinically relevant view of mGluR5 function in mental health than the view on preclinical data alone. We will also review the current research data on mGluR5 along the Research Domain Criteria (RDoC). Firstly, we found evidence of abnormal glutamate activity related to the positive and negative valence systems, which would suggest that antagonistic mGluR5 intervention has prominent anti-addictive, anti-depressive and anxiolytic effects. Secondly, there is evidence that mGluR5 plays an important role in systems for social functioning and the response to social stress. Finally, mGluR5's important role in sleep homeostasis suggests that this glutamate receptor may play an important role in RDoC's arousal and modulatory systems domain. Glutamate was previously mostly investigated in non-human studies, however initial human clinical PET research now also supports the hypothesis that, by mediating brain excitability, neuroplasticity and social cognition, abnormal metabotropic glutamate activity might predispose individuals to a broad range of psychiatric problems.
Collapse
Affiliation(s)
- Sylvia Terbeck
- School of Psychology, Faculty of Health and Human Sciences, University of Plymouth Plymouth, UK
| | - Funda Akkus
- Division of Molecular Psychiatry, Translational Research Center, Psychiatric University Hospital, University of Bern Bern, Switzerland
| | | | - Gregor Hasler
- Division of Molecular Psychiatry, Translational Research Center, Psychiatric University Hospital, University of Bern Bern, Switzerland
| |
Collapse
|
29
|
Erickson CA, Ray B, Maloney B, Wink LK, Bowers K, Schaefer TL, McDougle CJ, Sokol DK, Lahiri DK. Impact of acamprosate on plasma amyloid-β precursor protein in youth: a pilot analysis in fragile X syndrome-associated and idiopathic autism spectrum disorder suggests a pharmacodynamic protein marker. J Psychiatr Res 2014; 59:220-8. [PMID: 25300441 PMCID: PMC4253657 DOI: 10.1016/j.jpsychires.2014.07.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/09/2014] [Accepted: 07/14/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Understanding of the pathophysiology of autism spectrum disorder (ASD) remains limited. Brain overgrowth has been hypothesized to be associated with the development of ASD. A derivative of amyloid-β precursor protein (APP), secreted APPα (sAPPα), has neuroproliferative effects and has been shown to be elevated in the plasma of persons with ASD compared to control subjects. Reduction in sAPPα holds promise as a novel molecular target of treatment in ASD. Research into the neurochemistry of ASD has repeatedly implicated excessive glutamatergic and deficient GABAergic neurotransmission in the disorder. With this in mind, acamprosate, a novel modulator of glutamate and GABA function, has been studied in ASD. No data is available on the impact of glutamate or GABA modulation on sAPPα function. METHODS Plasma APP derivative levels pre- and post-treatment with acamprosate were determined in two pilot studies involving youth with idiopathic and fragile X syndrome (FXS)-associated ASD. We additionally compared baseline APP derivative levels between youth with FXS-associated or idiopathic ASD. RESULTS Acamprosate use was associated with a significant reduction in plasma sAPP(total) and sAPPα levels but no change occurred in Aβ40 or Aβ42 levels in 15 youth with ASD (mean age: 11.1 years). Youth with FXS-associated ASD (n = 12) showed increased sAPPα processing compared to age-, gender- and IQ-match youth with idiopathic ASD (n = 11). CONCLUSIONS Plasma APP derivative analysis holds promise as a potential biomarker for use in ASD targeted treatment. Reduction in sAPP (total) and sAPPα may be a novel pharmacodynamic property of acamprosate. Future study is required to address limitations of the current study to determine if baseline APP derivative analysis may predict subgroups of persons with idiopathic or FXS-associated ASD who may respond best to acamprosate or to potentially other modulators of glutamate and/or GABA neurotransmission.
Collapse
Affiliation(s)
| | - Balmiki Ray
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan Maloney
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Logan K. Wink
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Katherine Bowers
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tori L. Schaefer
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christopher J. McDougle
- Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deborah K. Sokol
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K. Lahiri
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA,Corresponding Author: Debomoy K. Lahiri, Ph.D., Professor, Departments of Psychiatry and of Medical & Molecular Genetics, Indiana University School of Medicine, Institute of Psychiatric Research, Neuroscience Research Building, 320 West 15th Street, NB 200C, Indianapolis, IN 46202-2266, USA, Tel: (317) 274-2706; Fax: (317) 231-0200
| |
Collapse
|
30
|
Erickson CA, Wink LK, Early MC, Stiegelmeyer E, Mathieu-Frasier L, Patrick V, McDougle CJ. Brief report: Pilot single-blind placebo lead-in study of acamprosate in youth with autistic disorder. J Autism Dev Disord 2014; 44:981-7. [PMID: 24052275 DOI: 10.1007/s10803-013-1943-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
RATIONALE An excitatory/inhibitory (E:I) imbalance marked by enhanced glutamate and deficient gamma-aminobutyric acid (GABA) neurotransmission may contribute to the pathophysiology of autism spectrum disorders (ASD). OBJECTIVES We report on the first single-blind placebo lead-in trial of acamprosate, a drug with putative mechanisms restoring E:I imbalance, in twelve youth with ASD. MATERIALS AND METHODS We conducted a 12-week single-blind, placebo lead-in study of acamprosate in youth age 5-17 years with autistic disorder. RESULTS Six of nine subjects who received active drug treatment were deemed treatment responders (defined by a score at final visit of "very much improved" or "much improved" on the Clinical Global Impressions Improvement scale) and ≥25% improvement on the Aberrant Behavior Checklist Social Withdrawal subscale. CONCLUSION Future larger-scale dose finding studies of acamprosate in ASD may be warranted given this preliminary indication of benefit.
Collapse
Affiliation(s)
- Craig A Erickson
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA,
| | | | | | | | | | | | | |
Collapse
|
31
|
Lin SK. Pharmacological means of reducing human drug dependence: a selective and narrative review of the clinical literature. Br J Clin Pharmacol 2014; 77:242-52. [PMID: 23701272 DOI: 10.1111/bcp.12163] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 04/28/2013] [Indexed: 12/20/2022] Open
Abstract
Substance abuse or addictive disorder is a global problem. A greater understanding of the associated changes in brain pathophysiology supports the notion that pharmacological treatments are part of the necessary treatment options. Craving is a core symptom of addictive disorder. It refers to a strong desire to use drugs again either to re-experience positive effects or to diminish negative experiences. Currently there are a number of medicines that are effective in the treatment of addictive disorders. These medications can either be for substitution (same pharmacological effect as the abused substance) or anticraving (decrease the craving of the abused substance). In this MEDLNE based review, specific compounds (naltrexone, acamprosate, topiramate, disulfiram, baclofen, N-acetylcysteine and bupropion) were selected that are known to diminish desire to use (anticraving effect) and that have been trialled for a number of different substance addictive disorders. Their therapeutic potential in clinical practice is discussed in light of their efficacy.
Collapse
Affiliation(s)
- Shih-Ku Lin
- Department of Psychiatry, Taipei City Hospital and Psychiatric Center, Taipei, Taiwan; Department of Psychiatry, School of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Rojas DC. The role of glutamate and its receptors in autism and the use of glutamate receptor antagonists in treatment. J Neural Transm (Vienna) 2014; 121:891-905. [PMID: 24752754 PMCID: PMC4134390 DOI: 10.1007/s00702-014-1216-0] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 04/06/2014] [Indexed: 12/11/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the brain and may be a key neurotransmitter involved in autism. Literature pertaining to glutamate and autism or related disorders (e.g., Fragile X syndrome) is reviewed in this article. Interest in glutamatergic dysfunction in autism is high due to increasing convergent evidence implicating the system in the disorder from peripheral biomarkers, neuroimaging, protein expression, genetics and animal models. Currently, there are no pharmaceutical interventions approved for autism that address glutamate deficits in the disorder. New treatments related to glutamatergic neurotransmission, however, are emerging. In addition, older glutamate-modulating medications with approved indications for use in other disorders are being investigated for re-tasking as treatments for autism. This review presents evidence in support of glutamate abnormalities in autism and the potential for translation into new treatments for the disorder.
Collapse
Affiliation(s)
- Donald C Rojas
- Department of Psychology, Campus Delivery 1876, Colorado State University, Fort Collins, CO, 80523, USA,
| |
Collapse
|
33
|
Abstract
Autism spectrum disorder is often comorbid with behavioral disturbances such as irritability, aggression and hyperactivity. Throughout the mid 2000s, several large-scale controlled clinical trials were published leading to the approval of two medications (aripiprazole and risperidone) for treatment of irritability in this condition. This review serves as an update regarding new research findings regarding psychopharmacology for children and adolescents with ASD. In summary, the past five years have yielded no further approved medications with ASD as a primary indication. Important new research results include 1) long-term safety and efficacy data (52 week) regarding treatment with aripiprazole for irritability, 2) consensus regarding potential harm from SSRIs for treatment of repetitive behaviors in children/ adolescents with ASD, 3) a randomized controlled trial showing modest benefits from atomoxetine on hyperactivity, 4) many novel agents currently under investigation.
Collapse
|
34
|
Kleijer KTE, Schmeisser MJ, Krueger DD, Boeckers TM, Scheiffele P, Bourgeron T, Brose N, Burbach JPH. Neurobiology of autism gene products: towards pathogenesis and drug targets. Psychopharmacology (Berl) 2014; 231:1037-62. [PMID: 24419271 DOI: 10.1007/s00213-013-3403-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 12/14/2013] [Indexed: 12/22/2022]
Abstract
RATIONALE The genetic heterogeneity of autism spectrum disorders (ASDs) is enormous, and the neurobiology of proteins encoded by genes associated with ASD is very diverse. Revealing the mechanisms on which different neurobiological pathways in ASD pathogenesis converge may lead to the identification of drug targets. OBJECTIVE The main objective is firstly to outline the main molecular networks and neuronal mechanisms in which ASD gene products participate and secondly to answer the question how these converge. Finally, we aim to pinpoint drug targets within these mechanisms. METHOD Literature review of the neurobiological properties of ASD gene products with a special focus on the developmental consequences of genetic defects and the possibility to reverse these by genetic or pharmacological interventions. RESULTS The regulation of activity-dependent protein synthesis appears central in the pathogenesis of ASD. Through sequential consequences for axodendritic function, neuronal disabilities arise expressed as behavioral abnormalities and autistic symptoms in ASD patients. Several known ASD gene products have their effect on this central process by affecting protein synthesis intrinsically, e.g., through enhancing the mammalian target of rapamycin (mTOR) signal transduction pathway or through impairing synaptic function in general. These are interrelated processes and can be targeted by compounds from various directions: inhibition of protein synthesis through Lovastatin, mTOR inhibition using rapamycin, or mGluR-related modulation of synaptic activity. CONCLUSIONS ASD gene products may all feed into a central process of translational control that is important for adequate glutamatergic regulation of dendritic properties. This process can be modulated by available compounds but may also be targeted by yet unexplored routes.
Collapse
Affiliation(s)
- Kristel T E Kleijer
- Department Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3984 CG, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Uzunova G, Hollander E, Shepherd J. The role of ionotropic glutamate receptors in childhood neurodevelopmental disorders: autism spectrum disorders and fragile x syndrome. Curr Neuropharmacol 2014; 12:71-98. [PMID: 24533017 PMCID: PMC3915351 DOI: 10.2174/1570159x113116660046] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/20/2013] [Accepted: 09/25/2013] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) and Fragile X syndrome (FXS) are relatively common childhood neurodevelopmental disorders with increasing incidence in recent years. They are currently accepted as disorders of the synapse with alterations in different forms of synaptic communication and neuronal network connectivity. The major excitatory neurotransmitter system in brain, the glutamatergic system, is implicated in learning and memory, synaptic plasticity, neuronal development. While much attention is attributed to the role of metabotropic glutamate receptors in ASD and FXS, studies indicate that the ionotropic glutamate receptors (iGluRs) and their regulatory proteins are also altered in several brain regions. Role of iGluRs in the neurobiology of ASD and FXS is supported by a weight of evidence that ranges from human genetics to in vitro cultured neurons. In this review we will discuss clinical, molecular, cellular and functional changes in NMDA, AMPA and kainate receptors and the synaptic proteins that regulate them in the context of ASD and FXS. We will also discuss the significance for the development of translational biomarkers and treatments for the core symptoms of ASD and FXS.
Collapse
Affiliation(s)
- Genoveva Uzunova
- Autism and Obsessive Compulsive Spectrum Program, Department of Psychiatry, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th St, Bronx, New York 10467-2490
| | - Eric Hollander
- Autism and Obsessive Compulsive Spectrum Program, Department of Psychiatry, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th St, Bronx, New York 10467-2490
| | - Jason Shepherd
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, 531A Wintrobe, 20N 1900 E, Salt Lake City, Utah 84132
| |
Collapse
|
36
|
Doll CA, Broadie K. Impaired activity-dependent neural circuit assembly and refinement in autism spectrum disorder genetic models. Front Cell Neurosci 2014; 8:30. [PMID: 24570656 PMCID: PMC3916725 DOI: 10.3389/fncel.2014.00030] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/21/2014] [Indexed: 01/23/2023] Open
Abstract
Early-use activity during circuit-specific critical periods refines brain circuitry by the coupled processes of eliminating inappropriate synapses and strengthening maintained synapses. We theorize these activity-dependent (A-D) developmental processes are specifically impaired in autism spectrum disorders (ASDs). ASD genetic models in both mouse and Drosophila have pioneered our insights into normal A-D neural circuit assembly and consolidation, and how these developmental mechanisms go awry in specific genetic conditions. The monogenic fragile X syndrome (FXS), a common cause of heritable ASD and intellectual disability, has been particularly well linked to defects in A-D critical period processes. The fragile X mental retardation protein (FMRP) is positively activity-regulated in expression and function, in turn regulates excitability and activity in a negative feedback loop, and appears to be required for the A-D remodeling of synaptic connectivity during early-use critical periods. The Drosophila FXS model has been shown to functionally conserve the roles of human FMRP in synaptogenesis, and has been centrally important in generating our current mechanistic understanding of the FXS disease state. Recent advances in Drosophila optogenetics, transgenic calcium reporters, highly-targeted transgenic drivers for individually-identified neurons, and a vastly improved connectome of the brain are now being combined to provide unparalleled opportunities to both manipulate and monitor A-D processes during critical period brain development in defined neural circuits. The field is now poised to exploit this new Drosophila transgenic toolbox for the systematic dissection of A-D mechanisms in normal versus ASD brain development, particularly utilizing the well-established Drosophila FXS disease model.
Collapse
Affiliation(s)
- Caleb A Doll
- Department of Biological Sciences, Vanderbilt University Nashville, TN, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University Nashville, TN, USA ; Kennedy Center for Research on Human Development, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
37
|
Baribeau DA, Anagnostou E. Social Communication is an Emerging Target for Pharmacotherapy in Autism Spectrum Disorder - A Review of the Literature on Potential Agents. JOURNAL OF THE CANADIAN ACADEMY OF CHILD AND ADOLESCENT PSYCHIATRY = JOURNAL DE L'ACADEMIE CANADIENNE DE PSYCHIATRIE DE L'ENFANT ET DE L'ADOLESCENT 2014; 23:20-30. [PMID: 24516474 PMCID: PMC3917666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 10/21/2013] [Indexed: 06/03/2023]
Abstract
OBJECTIVE To review the published literature and registered clinical trials on pharmacologic interventions targeting social communication impairment in Autism Spectrum Disorder (ASD). METHODS A comprehensive search of several databases (PubMed, MEDLINE, PsycINFO, Clinical trials.gov) was conducted to identify pharmacologic agents that have been, or will be, tested as treatments for social communication impairment in individuals with ASD. Evidence from basic science research supporting rational drug discovery is surveyed. RESULTS Data from animal models and early clinical trials suggest that novel and existing compounds, including N-methyl-D-aspartate (NMDA) modulators, γ-aminobutyric acid (GABA) agonists, metabotropic glutamate receptor (mGluR) antagonists and neuropeptides, may enhance social communication/function in ASD. Results from numerous Phase 2 and Phase 3 clinical trials are expected in the near future. CONCLUSIONS Recent evidence suggests that social communication may be an appropriate target for pharmacologic manipulation. It is hoped that, in combination with behavioural interventions, novel therapeutics may soon be clinically available to help improve social outcomes.
Collapse
Affiliation(s)
| | - Evdokia Anagnostou
- University of Toronto, Department of Pediatrics, Bloorview Research Institute, Toronto, Ontario
| |
Collapse
|
38
|
Sung M, Chin CH, Lim CG, Liew HSA, Lim CS, Kashala E, Weng SJ. What's in the pipeline? Drugs in development for autism spectrum disorder. Neuropsychiatr Dis Treat 2014; 10:371-81. [PMID: 24591832 PMCID: PMC3934669 DOI: 10.2147/ndt.s39516] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder with both core symptoms and associated symptoms (eg, irritability, aggression, and comorbidities) that affect both the individual and the family/systems around them. There have been recent advances in the understanding of the underlying pathophysiology of ASD pertaining to genetics, epigenetics, neurological, hormonal, and environmental factors that contribute to the difficulties found in individuals with ASD. With this improved understanding, there has been a shift in the application of psychopharmacology in ASD and its related disorders. A literature review was conducted to examine research published in the last 5 years between different classes of psychotropic medications and ASD. The broad scope of the existing literature for the use of conventional medications is summarized and novel medications are discussed.
Collapse
Affiliation(s)
- Min Sung
- Department of Child and Adolescent Psychiatry, Institute of Mental Health, Singapore
| | - Chee Hon Chin
- Department of Child and Adolescent Psychiatry, Institute of Mental Health, Singapore
| | - Choon Guan Lim
- Department of Child and Adolescent Psychiatry, Institute of Mental Health, Singapore
| | - Hwee Sen Alvin Liew
- Department of Child and Adolescent Psychiatry, Institute of Mental Health, Singapore
| | - Chau Sian Lim
- Department of Psychological Medicine, Khoo Teck Puat Hospital, Singapore
| | - Espérance Kashala
- Department of Child and Adolescent Psychiatry, Institute of Mental Health, Singapore
| | - Shih-Jen Weng
- Department of Child and Adolescent Psychiatry, Institute of Mental Health, Singapore
| |
Collapse
|
39
|
Farmer C, Thurm A, Grant P. Pharmacotherapy for the core symptoms in autistic disorder: current status of the research. Drugs 2013; 73:303-14. [PMID: 23504356 DOI: 10.1007/s40265-013-0021-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The current review covers extant literature on pharmacotherapy for core symptoms of autism. The core symptoms of autism include impairments in social interaction and communication, as well as the presence of restricted and repetitive behaviors. There are no known efficacious treatments for the core social symptoms, although effects on repetitive behaviors are indicated with some data. While studies of fenfluramine, secretin, opiates, and mood stabilizers generally find no effect, mixed results suggest more research is needed on antidepressants and atypical antipsychotics. Newer lines of research, including cholinergic and glutamatergic agents and oxytocin, will be of considerable interest in the future. However, research on the treatment of core symptoms is plagued by limitations in study design, statistical power, and other issues inherent to the study of treatments for autism (e.g., heterogeneity of the disorder) that continue to prevent the elucidation of efficacious treatments.
Collapse
Affiliation(s)
- Cristan Farmer
- Pediatrics and Developmental Neuroscience Branch, National Institute of Mental Health, 10 Center Drive MSC 1255, Building 10, Room 1C250, Bethesda, MD, 20892-1255, USA.
| | | | | |
Collapse
|
40
|
Erickson CA, Wink LK, Ray B, Early MC, Stiegelmeyer E, Mathieu-Frasier L, Patrick V, Lahiri DK, McDougle CJ. Impact of acamprosate on behavior and brain-derived neurotrophic factor: an open-label study in youth with fragile X syndrome. Psychopharmacology (Berl) 2013; 228:75-84. [PMID: 23436129 DOI: 10.1007/s00213-013-3022-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/29/2013] [Indexed: 12/13/2022]
Abstract
RATIONALE Fragile X syndrome (FXS) is an inherited form of developmental disability and a single gene cause of autism. As a disorder with increasingly understood pathophysiology, FXS is a model form of developmental disability for targeted drug development efforts. Preclinical animal model findings have focused targeted drug treatment development in FXS on an imbalance between excessive glutamate and deficient gamma-aminobutyric acid (GABA) neurotransmission. METHODS We conducted a prospective open-label 10-week trial of acamprosate in 12 youth aged 6-17 years (mean age: 11.9 years) with FXS. RESULTS Acamprosate use (mean dose: 1,054 ± 422 mg/day) was associated with treatment response (defined by a Clinical Global Impressions Improvement (CGI-I) scale score of "very much improved" or "much improved") in nine of 12 (75 %) subjects. Improvement was noted in social behavior and inattention/hyperactivity using multiple standard behavioral outcome measures. No significant adverse effects or changes in vital signs, including weight or laboratory measures, occurred during treatment with acamprosate. Additionally, pre- and post-treatment blood biomarker analyses looking at brain-derived neurotrophic factor (BDNF) levels found a significant increase in BDNF with treatment. In our pilot sample, treatment response did not correlate with change in BDNF with treatment. CONCLUSIONS Acamprosate was generally safe and well tolerated and was associated with a significant improvement in social behavior and a reduction in inattention/hyperactivity. The increase in BDNF that occurred with treatment may be a useful pharmacodynamic marker in future acamprosate studies. Given these findings, a double-blind, placebo-controlled study of acamprosate in youth with FXS is warranted.
Collapse
Affiliation(s)
- Craig A Erickson
- Department of Psychiatry, Indiana University School of Medicine, and James Whitcomb Riley Hospital for Children, Indianapolis, IN, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Brown MS, Singel D, Hepburn S, Rojas DC. Increased glutamate concentration in the auditory cortex of persons with autism and first-degree relatives: a (1)H-MRS study. Autism Res 2012; 6:1-10. [PMID: 23166003 DOI: 10.1002/aur.1260] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 09/26/2012] [Indexed: 11/06/2022]
Abstract
Increased glutamate levels have been reported in the hippocampal and frontal regions of persons with autism using proton magnetic resonance spectroscopy ((1)H-MRS). Although autism spectrum disorders (ASDs) are highly heritable, MRS studies have not included relatives of persons with ASD. We therefore conducted a study to determine if glutamate levels are elevated in people with autism and parents of children with autism. Single-voxel, point-resolved spectroscopy data were acquired at 3T for left and right hemisphere auditory cortical voxels in 13 adults with autism, 15 parents of children with autism, and 15 adult control subjects. The primary measure was glutamate + glutamine (Glx). Additional measures included n-acetyl-aspartate (NAA), choline (Cho), myoinositol (mI), and creatine (Cr). The autism group had significantly higher Glx, NAA, and Cr concentrations than the control subjects. Parents did not differ from control subjects on any measures. No significant differences in Cho or mI levels were seen among groups. No reliable correlations between autism symptom measures, and MRS variables were seen after Bonferroni correction for multiple comparisons. The elevation in Glx in autism is consistent with prior MRS data in the hippocampus and frontal lobe and may suggest increased cortical excitability. Increased NAA and Cr may indicate brain metabolism disturbances in autism. In the current study, we found no reliable evidence of a familial effect for any spectroscopy measure. This may indicate that these metabolites have no heritable component in autism, the presence of a compensatory factor in parents, or sample-specific limitations such as the participation of singleton families.
Collapse
Affiliation(s)
- Mark S Brown
- Department of Radiology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
42
|
Coghlan S, Horder J, Inkster B, Mendez MA, Murphy DG, Nutt DJ. GABA system dysfunction in autism and related disorders: from synapse to symptoms. Neurosci Biobehav Rev 2012; 36:2044-55. [PMID: 22841562 DOI: 10.1016/j.neubiorev.2012.07.005] [Citation(s) in RCA: 323] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 07/10/2012] [Accepted: 07/16/2012] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental syndromes characterised by repetitive behaviours and restricted interests, impairments in social behaviour and relations, and in language and communication. These symptoms are also observed in a number of developmental disorders of known origin, including Fragile X Syndrome, Rett Syndrome, and Foetal Anticonvulsant Syndrome. While these conditions have diverse etiologies, and poorly understood pathologies, emerging evidence suggests that they may all be linked to dysfunction in particular aspects of GABAergic inhibitory signalling in the brain. We review evidence from genetics, molecular neurobiology and systems neuroscience relating to the role of GABA in these conditions. We conclude by discussing how these deficits may relate to the specific symptoms observed.
Collapse
Affiliation(s)
- Suzanne Coghlan
- King's College London, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, United Kingdom
| | | | | | | | | | | |
Collapse
|